[1]
Musso, D.; Roche, C.; Robin, E.; Nhan, T.; Teissier, A.; Cao-Lormeau, V-M. Potential sexual transmission of Zika virus. Emerg. Infect. Dis., 2015, 21(2), 359.
[2]
Macnamara, F. Zika virus: A report on three cases of human infection during an epidemic of jaundice in Nigeria. Trans. R. Soc. Trop. Med. Hyg., 1954, 48(2), 139-145.
[3]
Besnard, M.; Lastere, S.; Teissier, A.; Cao-Lormeau, V.; Musso, D. Evidence of perinatal transmission of Zika virus, French Polynesia, December 2013 and February 2014. Euro Surveill., 2014, 19(13), 20751.
[4]
Musso, D.; Nhan, T.; Robin, E.; Roche, C.; Bierlaire, D.; Zisou, K.; Shan, Yan A.; Cao-Lormeau, V.; Broult, J. Potential for Zika virus transmission through blood transfusion demonstrated during an outbreak in French Polynesia, November 2013 to February 2014. Euro Surveill., 2014, 19(14), 20761.
[5]
Faizan, M.I.; Abdullah, M.; Ali, S.; Naqvi, I.H.; Ahmed, A.; Parveen, S. Zika virus-induced microcephaly and its possible molecular mechanism. Intervirology, 2016, 59(3), 152-158.
[6]
Dos Santos, T.; Rodriguez, A.; Almiron, M.; Sanhueza, A.; Ramon, P.; de Oliveira, W.K.; Coelho, G.E.; Badaró, R.; Cortez, J.; Ospina, M. Zika virus and the Guillain-Barré syndrome-case series from seven countries. N. Engl. J. Med., 2016, 375(16), 1598-1601.
[8]
Sirohi, D.; Chen, Z.; Sun, L.; Klose, T.; Pierson, T.C.; Rossmann, M.G.; Kuhn, R.J. The 3.8 Å resolution cryo-EM structure of Zika virus. Science, 2016, 352(6284), 467-470.
[9]
Baronti, C.; Piorkowski, G.; Charrel, R.N.; Boubis, L.; Leparc-Goffart, I.; de Lamballerie, X. Complete coding sequence of zika virus from a French polynesia outbreak in 2013. Genome Announc., 2014, 2(3), e00500-e00514.
[10]
Jain, R.; Coloma, J.; García-Sastre, A.; Aggarwal, A.K. Structure of the NS3 helicase from Zika virus. Nat. Struct. Mol. Biol., 2016, 23(8), 752-754.
[11]
Bollati, M.; Alvarez, K.; Assenberg, R.; Baronti, C.; Canard, B.; Cook, S.; Coutard, B.; Decroly, E.; de Lamballerie, X.; Gould, E.A. Structure and functionality in flavivirus NS-proteins: Perspectives for drug design. Antiviral Res., 2010, 87(2), 125-148.
[12]
Amberg, S.M.; Nestorowicz, A.; McCourt, D.W.; Rice, C.M. NS2B-3 proteinase-mediated processing in the yellow fever virus structural region: In vitro and in vivo studies. J. Virol., 1994, 68(6), 3794-3802.
[13]
Yamshchikov, V.F.; Compans, R.W. Processing of the intracellular form of the west Nile virus capsid protein by the viral NS2B-NS3 protease: An in vitro study. J. Virol., 1994, 68(9), 5765-5771.
[14]
Chambers, T.J.; Weir, R.C.; Grakoui, A.; McCourt, D.W.; Bazan, J.F.; Fletterick, R.J.; Rice, C.M. Evidence that the N-terminal domain of nonstructural protein NS3 from yellow fever virus is a serine protease responsible for site-specific cleavages in the viral polyprotein. Proc. Natl. Acad. Sci. USA, 1990, 87(22), 8898-8902.
[15]
Tian, H.; Ji, X.; Yang, X.; Xie, W.; Yang, K.; Chen, C.; Wu, C.; Chi, H.; Mu, Z.; Wang, Z. The crystal structure of Zika virus helicase: Basis for antiviral drug design. Protein Cell, 2016, 7(6), 450-454.
[16]
Cao, X.; Li, Y.; Jin, X.; Li, Y.; Guo, F.; Jin, T. Molecular mechanism of divalent-metal-induced activation of NS3 helicase and insights into Zika virus inhibitor design. Nucleic Acids Res., 2016, 44(21), 10505-10514.
[17]
Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The Protein Data Bank,
1999. In: International Tables for Crystallography Volume F: Crystallography
of biological macromolecules; Springer:, 2006; p. 675-
684.
[18]
Kuhn, R.J.; Zhang, W.; Rossmann, M.G.; Pletnev, S.V.; Corver, J.; Lenches, E.; Jones, C.T.; Mukhopadhyay, S.; Chipman, P.R.; Strauss, E.G. Structure of dengue virus: Implications for flavivirus organization, maturation, and fusion. Cell, 2002, 108(5), 717-725.
[19]
Zhang, X.; Ge, P.; Yu, X.; Brannan, J.M.; Bi, G.; Zhang, Q.; Schein, S.; Zhou, Z.H. Cryo-EM structure of the mature dengue virus at 3.5-Å resolution. Nat. Struct. Mol. Biol., 2013, 20(1), 105-110.
[20]
Narayana, K.R.; Reddy, M.S.; Chaluvadi, M.; Krishna, D. Bioflavonoids classification, pharmacological, biochemical effects and therapeutic potential. Indian J. Pharmacol., 2001, 33(1), 2-16.
[21]
Zandi, K.; Teoh, B-T.; Sam, S-S.; Wong, P-F.; Mustafa, M.R.; AbuBakar, S. Antiviral activity of four types of bioflavonoid against dengue virus type-2. Virol. J., 2011, 8(1), 560.
[22]
Kiat, T.S.; Pippen, R.; Yusof, R.; Ibrahim, H.; Khalid, N.; Rahman, N.A. Inhibitory activity of cyclohexenyl chalcone derivatives and flavonoids of fingerroot, Boesenbergia rotunda(L.), towards dengue-2 virus NS3 protease. Bioorg. Med. Chem. Lett., 2006, 16(12), 3337-3340.
[23]
Tomlinson, S.M.; Malmstrom, R.D.; Russo, A.; Mueller, N.; Pang, Y-P.; Watowich, S.J. Structure-based discovery of dengue virus protease inhibitors. Antiviral Res., 2009, 82(3), 110-114.
[24]
Keivan, Z.; Teoh, B-T.; Sam, S-S.; Wong, P-F.; Mustafa, M.R.; AbuBakar, S. In vitro antiviral activity of fisetin, rutin and naringenin against dengue virus type-2. J. Med. Plants Res., 2011, 5(23), 5534-5539.
[25]
Sanchez, I.; Gómez‐Garibay, F.; Taboada, J.; Ruiz, B. Antiviral effect of flavonoids on the dengue virus. Phytother. Res., 2000, 14(2), 89-92.
[26]
Chiow, K.; Phoon, M.; Putti, T.; Tan, B.K.; Chow, V.T. Evaluation of antiviral activities of Houttuynia cordata Thunb. Extract, quercetin, quercetrin and cinanserin on murine coronavirus and dengue virus infection. Asian Pac. J. Trop. Med., 2016, 9(1), 1-7.
[27]
Kim, S.; Thiessen, P.A.; Bolton, E.E.; Chen, J.; Fu, G.; Gindulyte, A.; Han, L.; He, J.; He, S.; Shoemaker, B.A. PubChem substance and compound databases. Nucleic Acids Res., 2015, 44(D1), D1202-D1213.
[28]
Irwin, J.J.; Sterling, T.; Mysinger, M.M.; Bolstad, E.S.; Coleman, R.G. ZINC: A free tool to discover chemistry for biology. J. Chem. Inf. Model., 2012, 52(7), 1757-1768.
[29]
Basavannacharya, C.; Vasudevan, S.G. Suramin inhibits helicase activity of NS3 protein of dengue virus in a fluorescence-based high throughput assay format. Biochem. Biophys. Res. Commun., 2014, 453(3), 539-544.
[30]
Mastrangelo, E.; Pezzullo, M.; De Burghgraeve, T.; Kaptein, S.; Pastorino, B.; Dallmeier, K.; de Lamballerie, X.; Neyts, J.; Hanson, A.M.; Frick, D.N. Ivermectin is a potent inhibitor of flavivirus replication specifically targeting NS3 helicase activity: New prospects for an old drug. J. Antimicrob. Chemother., 2012, 67(8), 1884-1894.
[31]
de Wispelaere, M.; LaCroix, A.J.; Yang, P.L. The small molecules AZD0530 and dasatinib inhibit dengue virus RNA replication via Fyn kinase. J. Virol., 2013, 87(13), 7367-7381.
[32]
Grosdidier, A.; Zoete, V.; Michielin, O. SwissDock, a protein-small molecule docking web service based on EADock DSS. Nucleic
Acids Res, 2011, 39(suppl_2), W270-W277.
[33]
Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem., 2010, 31(2), 455-461.
[34]
Alam, A.; Tamkeen, N.; Imam, N.; Farooqui, A.; Ahmed, M.M.; Tazyeen, S.; Ali, S.; Malik, M.Z.; Ishrat, R. Pharmacokinetic and molecular docking studies of plant-derived natural compounds to exploring potential anti-alzheimer activity.In Silico Approach for Sustainable Agriculture; Springer, 2018, pp. 217-238.
[35]
Arup, K. Ghose, Vellarkad N. Viswanadhan, John J. Wendoloski. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J. Comb. Chem., 1999, 1(1), 55-68.
[36]
Seeliger, D.; de Groot, B.L. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J. Comput. Aided Mol. Des., 2010, 24(5), 417-422.
[37]
DeLano, W.L. The PyMOL molecular graphics system; Palo Alto, CA, USA, 2002.
[38]
Wallace, A.C.; Laskowski, R.A.; Thornton, J.M. LIGPLOT: A program to generate schematic diagrams of protein-ligand interactions. Protein Eng. Des. Sel., 1995, 8(2), 127-134.
[39]
Ahsan, M.J.; Samy, J.G.; Khalilullah, H.; Nomani, M.S.; Saraswat, P.; Gaur, R.; Singh, A. Molecular properties prediction and synthesis of novel 1,3,4-oxadiazole analogues as potent antimicrobial and antitubercular agents. Bioorg. Med. Chem. Lett., 2011, 21(24), 7246-7250.
[40]
Bavan, S.; Sherman, B.; Luetje, C.W.; Abaffy, T. Discovery of novel ligands for mouse olfactory receptor MOR42-3 using an in silico screening approach and in vitro validation. PLoS One, 2014, 9(3), e92064.
[41]
Gruba, N.; Rodriguez Martinez, J.I.; Grzywa, R.; Wysocka, M.; Skoreński, M.; Burmistrz, M.; Łęcka, M.; Lesner, A.; Sieńczyk, M.; Pyrć, K. Substrate profiling of Zika virus NS2B‐NS3 protease. FEBS Lett., 2016, 590(20), 3459-3468.
[42]
Tian, H.; Ji, X.; Yang, X.; Zhang, Z.; Lu, Z.; Yang, K.; Chen, C.; Zhao, Q.; Chi, H.; Mu, Z. Structural basis of Zika virus helicase in recognizing its substrates. Protein Cell, 2016, 7(8), 562-570.
[43]
Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings1. Adv. Drug Deliv. Rev., 2001, 46(1-3), 3-26.
[44]
van de Waterbeemd, H.; Camenisch, G.; Folkers, G.; Chretien, J.R.; Raevsky, O.A. Estimation of blood-brain barrier crossing of drugs using molecular size and shape, and H-bonding descriptors. J. Drug Target., 1998, 6(2), 151-165.
[45]
Veber, D.F.; Johnson, S.R.; Cheng, H-Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem., 2002, 45(12), 2615-2623.
[46]
Klages, J.; Coles, M.; Kessler, H. NMR-based screening: A powerful tool in fragment-based drug discovery. Analyst, 2007, 132(7), 692-705.
[47]
Blake, J.F. Examination of the computed molecular properties of compounds selected for clinical development. Biotechniques, 2003, 34, S16-S20.
[48]
Abdullah, M.; Sher Ali, A.T.; Naqvi, I.H.; Verma, H.N.; Ahmed, A.; Kazim, S.N.; Parveen, S. Zika viral infection and its future prospects. Indian J. Health Sci. Care, 2016, 3(2), 78-84.
[49]
Badshah, S.L.; Naeem, A.; Mabkhot, Y. The new high resolution crystal structure of NS2B-NS3 protease of Zika virus. Viruses, 2017, 9(1), 7.
[50]
Li, Y.; Zhang, Z.; Phoo, W.W.; Loh, Y.R.; Wang, W.; Liu, S.; Chen, M.W.; Hung, A.W.; Keller, T.H.; Luo, D. Structural dynamics of Zika virus NS2B-NS3 protease binding to dipeptide inhibitors. Structure, 2017, 25(8), 1242-1250.
[51]
Lee, H.; Ren, J.; Nocadello, S.; Rice, A.J.; Ojeda, I.; Light, S.; Minasov, G.; Vargas, J.; Nagarathnam, D.; Anderson, W.F. Identification of novel small molecule inhibitors against NS2B/NS3 serine protease from Zika virus. Antiviral Res., 2017, 139, 49-58.
[52]
Mottin, M.; Braga, R.C.; da Silva, R.A.; da Silva, J.H.M.; Perryman, A.L.; Ekins, S.; Andrade, C.H. Molecular dynamics simulations of Zika virus NS3 helicase: Insights into RNA binding site activity. Biochem. Biophys. Res. Commun., 2017, 492(4), 643-651.
[53]
Xu, M.; Lee, E.M.; Wen, Z.; Cheng, Y.; Huang, W-K.; Qian, X.; Julia, T.; Kouznetsova, J.; Ogden, S.C.; Hammack, C. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat. Med., 2016, 22(10), 1101.
[54]
Andrews, K.T.; Fisher, G.; Skinner-Adams, T.S. Drug repurposing and human parasitic protozoan diseases. Int. J. Parasitol. Drugs Drug Resist., 2014, 4(2), 95-111.
[55]
Sanseau, P.; Koehler, J. Computational methods for drug repurposing; Oxford University Press, 2011.
[56]
Liu, S.; Ewing, M.; Anglard, P.; Trahan, E.; La Rocca, R.V.; Myers, C.E.; Linehan, W.M. The effect of suramin, tumor necrosis factor and interferon γ on human prostate carcinoma. J. Urol., 1991, 145(2), 389-392.
[57]
Wagstaff, K.M.; Sivakumaran, H.; Heaton, S.M.; Harrich, D.; Jans, D.A. Ivermectin is a specific inhibitor of importin α/β-mediated nuclear import able to inhibit replication of HIV-1 and dengue virus. Biochem. J., 2012, 443(3), 851-856.
[58]
Campbell, W.C. Ivermectin and abamectin; Springer Science & Business Media, 2012.
[59]
Gratacap, M-P.; Martin, V.; Valéra, M-C.; Allart, S.; Garcia, C.; Sié, P.; Recher, C.; Payrastre, B. The new tyrosine-kinase inhibitor and anticancer drug dasatinib reversibly affects platelet activation in vitro and in vivo. Blood, 2009, 114(9), 1884-1892.
[60]
Tjioe, K.C.; Tostes Oliveira, D.; Gavard, J. luteolin impacts on the dna damage pathway in oral squamous cell carcinoma. Nutr. Cancer, 2016, 68(5), 838-847.